12
Progesterone During Pregnancy: Endocrine–Immune Cross Talk in Mammalian Species and the Role of Stress Petra Arck 1 , Peter J Hansen 2 , Biserka Mulac Jericevic 3 , Marie-Pierre Piccinni 4 , Julia Szekeres-Bartho 5 1 Charite ´ , University Medicine Berlin, Berlin, Germany; 2 Department of Animal Sciences, University of Florida, Gainesville, FL, USA; 3 Department of Physiology and Immunology, Medical School, University of Rijeka, Rijeka, Croatia; 4 Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Department of Internal Medicine – Immunoallergology unit – viale Morgagni, Florence, Italy; 5 Department of Medical Microbiology and Immunology Medical School, Pecs University, Pecs, Hungary Introduction Pregnancy is characterized by overall hormonal changes. Progesterone (P) is critical for not only the establishment but also for the maintenance of preg- nancy, as its functions support ovulation and uterine as well as mammary gland development. Compared to the low levels (1–2 nmol L) during the follicu- lar phase of the menstrual cycle P concentrations increase to 15–20, 35–50, and 20–40 nmol L in the early- mid-, and late-luteal phases respectively. The major source of P during pregnancy is the cor- pus luteum of the ovary and, if pregnancy occurs, in many species, including humans and rodents, P production is eventually sustained by the pla- centa. 1 In humans, P production gradually rises dur- ing gestation to reach a level of 3 lg g of placental tissue (1–10 lm), whereas the serum concentrations of P range from about 100 to 500 nm during preg- nancy. 2 The need for P in maintaining pregnancy is shown by the fact that blocking of P binding sites causes abortion in human and also in various animal species. Besides its endocrine effects P acts as an ‘im- munosteroid’. Successful pregnancy depends on maternal tolerance of the fetal ‘semi-allograft’ (Clark et al., current issue). Here, P blocks very early T-cell Keywords NK cells, pregnancy, progesterone Correspondence Julia Szekeres Bartho, MD, PhD, Department of Medical Microbiology and Immunology, Medical School, Pecs University, 12 Szigeti Str., H-7643 Pecs, Hungary. E-mail: [email protected] Submitted June 16, 2007; accepted June 21, 2007. Citation Arck P, Hansen PJ, Mulac Jericevic B, Piccinni M-P, Szekeres-Bartho J. Progesterone during pregnancy: endocrine-immune cross talk in mammalian species and the role of stress. Am J Reprod Immunol 2007; 58:268–279 doi:10.1111/j.1600-0897.2007.00512.x Progesterone is critical for the establishment and the maintenance of pregnancy, both by its endocrine and immunological effects. The geno- mic actions of progesterone are mediated by the intracellular progester- one receptors; A and B. A protein called P-induced blocking factor (PIBF), by inducing a T H2 dominant cytokine production, mediates the immunological effects of progesterone. Progesterone plays a role in uter- ine homing of NK cells and up-regulates HLA-G gene expression, the ligand for various NK inhibitory receptors. At high concentrations pro- gesterone is a potent inducer of Th2-type cytokines as well as of LIF and M-CSF production by T cells. Though a key role for progesterone in cre- ating local immunosuppression has been conserved during the evolution of an epitheliochorial placenta, there has been some divergence in the pattern of endocrine-immunological cross talk in Bovidae. In sheep, uter- ine serpin, a progesterone-induced endometrial protein, mediates the immunosuppressive effects of progesterone. Epidemiological studies sug- gest the role of stress in premature pregnancy termination and exposure to stress induces abortion in mice via a significant reduction in progester- one levels, accompanied by reduced serum levels of PIBF. These effects are corrected by progesterone supplementation. These findings indicate the significance of a progesterone-dependent immuno-modulation in maternal tolerance of the fetus, which is discussed in this review. REVIEW ARTICLE American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors 268 Journal compilation ª 2007 Blackwell Munksgaard

Progesterone During Pregnancy: Endocrine–Immune Cross Talk ...animal.ifas.ufl.edu/hansen/publications_pdf/docs/2007_arck.pdf · Progesterone During Pregnancy: Endocrine–Immune

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Progesterone During Pregnancy: Endocrine–Immune Cross Talkin Mammalian Species and the Role of StressPetra Arck1, Peter J Hansen2, Biserka Mulac Jericevic3, Marie-Pierre Piccinni4, Julia Szekeres-Bartho5

    1Charité, University Medicine Berlin, Berlin, Germany;2Department of Animal Sciences, University of Florida, Gainesville, FL, USA;3Department of Physiology and Immunology, Medical School, University of Rijeka, Rijeka, Croatia;4Center of Excellence for Research, Transfer and High Education DENOTHE of the University of Florence, Department of Internal Medicine –

    Immunoallergology unit – viale Morgagni, Florence, Italy;5Department of Medical Microbiology and Immunology Medical School, Pecs University, Pecs, Hungary

    Introduction

    Pregnancy is characterized by overall hormonal

    changes. Progesterone (P) is critical for not only the

    establishment but also for the maintenance of preg-

    nancy, as its functions support ovulation and uterine

    as well as mammary gland development. Compared

    to the low levels (1–2 nmol ⁄ L) during the follicu-lar phase of the menstrual cycle P concentrations

    increase to 15–20, 35–50, and 20–40 nmol ⁄ L in theearly- mid-, and late-luteal phases respectively.

    The major source of P during pregnancy is the cor-

    pus luteum of the ovary and, if pregnancy occurs, in

    many species, including humans and rodents,

    P production is eventually sustained by the pla-

    centa.1 In humans, P production gradually rises dur-

    ing gestation to reach a level of 3 lg ⁄ g of placentaltissue (1–10 lm), whereas the serum concentrationsof P range from about 100 to 500 nm during preg-

    nancy.2 The need for P in maintaining pregnancy is

    shown by the fact that blocking of P binding sites

    causes abortion in human and also in various animal

    species. Besides its endocrine effects P acts as an ‘im-

    munosteroid’. Successful pregnancy depends on

    maternal tolerance of the fetal ‘semi-allograft’ (Clark

    et al., current issue). Here, P blocks very early T-cell

    Keywords

    NK cells, pregnancy, progesterone

    Correspondence

    Julia Szekeres Bartho, MD, PhD, Department

    of Medical Microbiology and Immunology,

    Medical School, Pecs University, 12 Szigeti

    Str., H-7643 Pecs, Hungary.

    E-mail: [email protected]

    Submitted June 16, 2007;

    accepted June 21, 2007.

    Citation

    Arck P, Hansen PJ, Mulac Jericevic B, Piccinni

    M-P, Szekeres-Bartho J. Progesterone during

    pregnancy: endocrine-immune cross talk in

    mammalian species and the role of stress. Am

    J Reprod Immunol 2007; 58:268–279

    doi:10.1111/j.1600-0897.2007.00512.x

    Progesterone is critical for the establishment and the maintenance of

    pregnancy, both by its endocrine and immunological effects. The geno-

    mic actions of progesterone are mediated by the intracellular progester-

    one receptors; A and B. A protein called P-induced blocking factor

    (PIBF), by inducing a TH2 dominant cytokine production, mediates the

    immunological effects of progesterone. Progesterone plays a role in uter-

    ine homing of NK cells and up-regulates HLA-G gene expression, the

    ligand for various NK inhibitory receptors. At high concentrations pro-

    gesterone is a potent inducer of Th2-type cytokines as well as of LIF and

    M-CSF production by T cells. Though a key role for progesterone in cre-

    ating local immunosuppression has been conserved during the evolution

    of an epitheliochorial placenta, there has been some divergence in the

    pattern of endocrine-immunological cross talk in Bovidae. In sheep, uter-

    ine serpin, a progesterone-induced endometrial protein, mediates the

    immunosuppressive effects of progesterone. Epidemiological studies sug-

    gest the role of stress in premature pregnancy termination and exposure

    to stress induces abortion in mice via a significant reduction in progester-

    one levels, accompanied by reduced serum levels of PIBF. These effects

    are corrected by progesterone supplementation. These findings indicate

    the significance of a progesterone-dependent immuno-modulation in

    maternal tolerance of the fetus, which is discussed in this review.

    REVIEW ARTICLE

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    268 Journal compilation ª 2007 Blackwell Munksgaard

  • lymphopoiesis during pregnancy3 and controls the

    bias towards a pregnancy protective immune

    milieu,4 which involves an immunomodulatory

    protein, known as P-induced blocking factor (PIBF).5

    Such observations might be just the ‘tip of the ice-

    berg’ of insights into the cross talk between the

    endocrine and the immune systems,6–8 the underly-

    ing mechanisms that operate throughout gestation

    are not completely understood.

    Progesterone, its receptors and mechanisms of

    actions

    Progesterone (P), one of the key players in the inter-

    action between the endocrine and immune systems,

    regulates menstrual bleeding, tissue repair and

    regeneration, inflammation, angiogenesis, blastocyst

    implantation, and maintenance of pregnancy. These

    events result from precisely coordinated activity of

    molecular pathways that ensure endometrial cell

    proliferation, differentiation, cell survival, leukocyte

    trafficking, apoptosis, and angiogenesis.

    Genomic and non-genomic pathways mediate the

    biological activities of P. The P regulated genomic

    pathway depends on two progesterone receptor

    (nPR) isoforms, PR-A and PR-B, both members of

    the nuclear receptor superfamily of transcription fac-

    tors.9,10 PR-A and PR-B are the products of the same

    gene, transcribed under control of two distinct pro-

    moters. The PR-A and PR-B isoforms differ, in that

    PR-B contains an additional N-terminal stretch of

    approximately 165 amino acids.

    In addition to regulation by P, the transcriptional

    activity of nPR isoforms can be regulated through

    alternation of expression level, interaction with co-

    activators and post-translational modification of

    nPR.9,10 Spatial and temporal expression of the PR-A

    and PR-B vary in reproductive tissues as a conse-

    quence of the developmental and the hormonal sta-

    tus as well as of carcinogenesis.

    In mice, null mutation of the nPR gene revealed

    that transcriptional activity of nPR controls the uter-

    ine immune environment as well as endometrial

    receptivity and decidualization.11,12 Also functionally

    active nPRs in the thymus are required for thymic

    involution during pregnancy and for a normal fertil-

    ity.13 Studies with mice in which PR-A and PR-B

    expression were selectively ablated demonstrate that

    PR-A and PR-B isoforms are functionally distinct

    transcription factors.13,14 Briefly, P-induced activa-

    tion of PR-A is both necessary and sufficient for the

    establishment and the maintenance of pregnancy,

    but elicits reduced pregnancy-stimulated mammary

    gland morphogenesis. In contrast, P-induced

    transcriptional activity of the PR-B isoform is insuffi-

    cient for implantation and the maintenance of preg-

    nancy, and mice lacking PR-A are infertile. These

    findings imply that the relative expression of the

    two isoforms is critical for the appropriate reproduc-

    tive tissue responses to P.15

    Although the genomic pathway of P action has

    been extensively studied, so far only a few P-regu-

    lated genes have been identified in the peri-implan-

    tation uterus. These genes include amphiregulin (a

    member of EGF superfamily), histidine decarboxyl-

    ase (an enzyme that converts histidine to histamine),

    Hox-A10 and Hox-A11 (both members of homeobox

    gene family), calcitonin (a peptide hormone), proen-

    cephalin (neuropeptide), immune response gene 1

    (Irg-1), MUC1 (a glycoprotein component of apical

    glycocalyx), indian hedgehog (mophogen), and

    galectin-1.16

    Hox-A10 deficiency in mice leads to severe local

    immunological disturbances, characterized by a poly-

    clonal proliferation of T cells that occurs in absence

    of the normal P-mediated immunosuppression in the

    peri-implantation uterus.17 Natural killer (NK) cell

    constitute the predominant leukocyte population

    present in endometrium at the time of implantation

    and early pregnancy (Santoni et al.18, current issue).

    Hox-A10 deficiency in mice alters region specific

    gene expression and compromises NK cell differenti-

    ation, but not trafficking of NK precursors cells dur-

    ing decidualization.19

    Estrogen-induced MUC1 is expressed by human

    villous syncytio- and cytotrophoblast as well as by

    invasive extravillous cytotrophoblast.20 It was pro-

    posed that blastocyst implantation is regulated by

    a uterine barrier, whereby a high density of MUC1

    at the epithelial cell surface can inhibit blastocyst

    adhesion.21 In mice MUC1 expression in the uterine

    epithelium is down-regulated during the window of

    implantation, and studies on genetically modified

    mice suggest that PR-A antagonizes Muc1 expression,

    which may subsequently allow blastocyst adhesion.22

    Indian hedgehog (Ihh) is another gene, regulated

    by P. Ihh plays a critical role in communication

    (required for embryo implantation) between the

    uterine epithelium and stroma.23 Ihh belongs to

    hedgehog family of morphogens that regulate cell

    proliferation, differentiation and cell–cell communi-

    cation, all of which may be involved in successful

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 269

  • decidualization and maintenance of fetal tolerance.

    Finally, galectin-1, which is also under the

    regulation of P, appears to be of importance, since its

    expression is down regulated on placental villous tis-

    sues from patients with spontaneous miscarriages.24

    Non-genomic actions of P include (a) P-induced

    acrosomal reaction, (b) P-induced resumption of

    meiosis, and (c) P induced decrease in neuronal

    excitability and anesthesia.25 The non-genomic

    actions are characterized by a fast response to

    P (latency of minutes rather than hours) and no

    requirement for de novo protein or RNA synthesis.

    Non-genomic actions of P appear to operate through

    membrane specific G protein-coupled receptors.

    Proper functional communication between the

    genomic and non-genomic P-regulated signaling

    pathways could be critical for the establishment of

    a correct endocrine-immune interaction in human

    endometrium during the establishment and mainte-

    nance of pregnancy.25

    Clearly, a profound knowledge of the key molecu-

    lar signals that are essential for the establishment of

    the receptive uterus will open therapeutic

    approaches in the development of new strategies for

    the treatment of implantation failures. Although

    technological advancement in functional genomics

    and proteomics allows identification of the differen-

    tially regulated genes during the implantation win-

    dow26,27 ethical considerations preclude an in-depth

    investigation of early molecular events in humans.

    Therefore, animal models, such as mouse knockout

    models will continue to be invaluable tools for

    studying the molecular events involved in the estab-

    lishment and maintenance of pregnancy.28

    The effect of P on the immune system

    With the exception of human leukocyte (HLA)-C,

    polymorphic major histocompatibility complex

    (MHC) is not expressed on human trophoblast, and

    this creates a unique immunological situation.

    Though decidual macrophages and dendritic cells

    can present fetal antigens to both decidual CD4+ and

    CD8+ cells, trophoblast-presented antigens are unli-

    kely to be recognized in an MHC-restricted fashion.

    In the decidua, there is a significantly increased

    number of activated c ⁄ d TCR-positive cells.29,30 Asmost c ⁄d T cells are capable to recognize unprocessedforeign antigens without MHC restriction, they could

    be candidates for ‘seeing’ trophoblast presented anti-

    gens. In peripheral blood of healthy pregnant

    women the number of c ⁄ d T cells is a significantlyincreased, and almost all of them express nPRs,31

    suggesting a prior activation. These cells could be of

    decidual origin, which, after activation by tropho-

    blast presented antigens, appear in peripheral circu-

    lation.

    Uterine dendritic cells (DC) have been proposed to

    serve as a switchboard between fetal rejection and

    tolerance.32–34 DCs are the most potent antigen pre-

    senting cells (APCs) involved in the innate immune

    response and in the maintenance of tolerance.35 The

    regulation of their maturation, migration, and

    expression of stimulatory and costimulatory mole-

    cules has major consequences on the immune

    response, whereby endogenous factors regulating DC

    function are poorly understood. Immature DCs exhi-

    bit a tolerogenic phenotype, characterized by low

    expression of costimulatory molecules (CD40, CD80,

    and CD86), low production of proinflammatory

    cytokines, increased production of IL-10, and capac-

    ity to induce regulatory T cells with suppressive

    actions, all of which will promote pregnancy mainte-

    nance. Immature DC reside in early pregnancy

    decidua in humans32,36 and mice34,37 and possibly

    serve as sentinel cells of the tissue environment for

    potential danger signals. However, in murine preg-

    nancies with high abortion rates, an increase of

    mature APC can be observed.34 By blocking crucial

    ligands required on APCs to induce T-cell activation,

    mechanisms of fetal tolerance are restored in aborton-

    prone pregnancies.38 P has been shown to inhibit

    mature dendritic cells as well as DC-stimulated pro-

    liferation of T cells in a receptor-mediated fashion.39

    The effect of P on the immune system of pregnant

    women could be partly receptor-mediated.40–43

    Recent finding suggest that P might act directly

    through membrane specific P receptors to suppress

    T-cell activation during pregnancy.44 Following

    recognition of fetal antigens, activated maternal c ⁄ dT cells express nPRs,31 and upon P binding, they pro-

    duce a mediator namely PIBF.5,45 In urine samples of

    healthy pregnant women, PIBF concentration contin-

    uously increases until the 37th week of gestation,

    followed by a slow decrease until term. In pregnancies

    that end up in miscarriage or pre-term delivery, uri-

    nary PIBF levels fail to increase during pregnancy.46

    By signaling via the Jak ⁄ STAT pathway,47 PIBFinduces a TH2 dominant cytokine production

    48 and

    in a cytokine-mediated way blocks NK activity.49

    Neutralization of endogenous PIBF activity in

    pregnant mice by specific anti-PIBF antibody causes

    ARCK ET AL.

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    270 Journal compilation ª 2007 Blackwell Munksgaard

  • a significant reduction in the number of viable

    fetuses, and this is associated with an increased sple-

    nic NK activity, together reduced IL-10 and

    increased interferon-c (IFN-c) production of thespleen cells.50 These are corrected by treatment of

    the pregnant animals with anti-NK antibodies,50

    suggesting that in mice PIBF contributes to the

    success of pregnancy and that the major part of its

    pregnancy-protective effect lies in controlling NK

    activity.

    In rodents and also in human hormonally

    controlled uterine NK cells play an important role in

    creating a suitable environment for the establish-

    ment of pregnancy.51 The temporal and spatial distri-

    bution of these cells suggests that one of the

    functions of these cells might be the control of

    placentation. Decidual NK cells secrete an array of

    angiogenic factors and induce vascular growth in the

    decidua. By producing interleukin-8 and interferon-

    inducible protein-10 chemokines decidual NK cells

    regulate trophoblast invasion.52

    Henderson et al.53 demonstrated the absence of PR

    in purified decidual NK cells, and thus a genomic

    action of P on these cells is unlikely. Nevertheless,

    P affects decidual NK cells in several ways.

    Van den Heuvel et al.42 reported that P plays

    a role in uterine homing of NK cells by promoting

    NK cell interactions with the endothelium. NK cell

    migration to the endometrium is also supported by

    sex-hormone-induced specific endometrial produc-

    tion of chemokines.54

    Decidual NK cells show a low spontaneous cyto-

    toxic activity in spite of their high perforin con-

    tent.55 Many of them express inhibitory receptors

    that recognize non-polymorphic MHC molecules.

    P has been shown to up-regulate HLA-G gene

    expression,56 and the increased availability of P pro-

    tects the trophoblast from NK-mediated killing.

    Based on their cytokine secretion profile human

    CD4+ T helper cells, can be subdivided into at least

    three distinct functional subsets.57,58

    Human type 1 (Th1) CD4+ T cells that produce

    interleukin (IL-2), tumor necrosis factor (TNF-a), andIFN-c are the main effectors of host defense againstinfections by intracellular parasites. On the other

    hand, human type 2 (Th2) CD4+ T cells produce IL-4,

    IL-5, IL-13 and IL-10, which together with IL-4 inhi-

    bit several macrophage functions. A third type (Th0)

    produces both Th1- and Th2-type cytokines. Recently

    additional CD4+ cell subsets have been identified:

    Th3 cells, which produce TGF-b and IL-10,59 Th17

    cells, which produce IL-17A and IL-22,60 and

    regulatory T cells (T reg) secreting IL-10 and TGF-b.61

    Cytokines produced by APCs and lymphocytes

    affect the development of Th1 and Th2 responses

    both in vitro and in vivo. IFN-c, IL-12, and IFN-aexert critical effects on CD4+ subset maturation by

    inducing Th1 expansion,62,63 while IL-4 is needed

    for Th2 cell maturation.62 Steroid hormones control

    the cytokine profile of T cells. Glucocorticoids and

    1,25-dihydroxy-vitamin D3 increase IL-4,64,65

    whereas dihydrotestosterone decreases IL-4 and IL-5

    production.66 The polypeptide hormone relaxin pre-

    dominantly produced by the corpus luteum and

    decidua during pregnancy favors the development of

    IFN-c-producing T cells.67 In two murine T-cell lines(NIMP-TH1 and EL4) dexamethasone negatively reg-

    ulates IL-5 gene expression, whereas testosterone

    and P induce the expression of IL-5 gene.68

    Progesterone also affects the differentiation of rest-

    ing peripheral blood T cells into Th1-, Th0-, or

    Th2-like clones and the production of IL-4 by

    human T-cell clones4 via the development of anti-

    gen-specific CD4+ T cell lines with an enhanced abil-

    ity to produce IL-4 and IL-5.4 In addition, P induces

    IL-4 mRNA expression and the production of detect-

    able amounts of IL-4 in Th1-type T-cell clones (able

    to produce IFN-c only without P).4 IL-4 productionby Th1 T-cell clones in response to P was associated

    with the expression of CD30 (a molecule preferen-

    tially expressed by IL4-producing T cells).4

    These results indicate that P at concentrations that

    are higher than those found in serum during preg-

    nancy, but comparable to those present at the

    feto-maternal interface,1 functions as a potent indu-

    cer of Th2-type cytokine production, which could be

    independently confirmed by others.2 Further, P pro-

    motes the development of LIF (leukemia inhibitory

    factor),69 as well as macrophage colony-stimulating

    factor (M-CSF)-producing T cells.70 Progesterone-

    induced LIF (essential for the embryo implantation)

    and M-CSF (important for pregnancy development)

    production was mediated by IL-4, produced by

    T cells in response to P. The effects of P were hor-

    mone-specific, as P receptors are found on activated

    T cells and P analogues (4 pregnen 20 b-ol 3- one, 4pregnen 20- a ol 3- one, and 5 pregnen 3 b- ol 20-one) have no effect on cytokine production by either

    T-cell lines and clones.4,40 The mechanisms by which

    P acts on T-cell differentiation are still unknown.

    It has been suggested that a Th1 to Th2 switch

    at the feto-maternal interface plays a role in the

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 271

  • maintenance of successful pregnancy.71 In line with

    this, placental IL-4 mRNA expression in mice was

    found to be 5- to 10-fold higher than in peripheral

    blood.72 Furthermore, defective IL-4 production by

    decidual CD4+ as well as CD8+ T cells, and defective

    of IL-10, LIF, and M-CSF production by decidual

    CD4+ T cells were detectable in women with unex-

    plained recurrent abortion at the time of miscar-

    riage.69 These data suggest that in human the

    success of pregnancy is associated with the produc-

    tion of Th2-type cytokines, LIF and M-CSF by T cells

    at materno-fetal interface. P, which, at concentra-

    tions comparable with those present at the materno-

    fetal interface during pregnancy, is not only a potent

    inducer of Th2-type cytokines (i.e. IL-4 and IL-5),4

    but also of LIF and M-CSF production by T cells69,70

    may be at least in part responsible for a Th2 switch

    at maternofetal interface.73 IL-4 produced by decid-

    ual Th2 cells can in turn promote the development

    of T cells producing LIF and M-CSF,69,70 which seem

    to be important for embryo implantation and devel-

    opment. Both IL-4 and IL-10 can inhibit the devel-

    opment and function of Th1 cells and macrophages,

    thus preventing the allograft rejection.

    These findings indicate that the immunological

    effects of P contribute to the complex network of

    regulatory pathways in the cause of fetal allograft

    survival.

    The effect of stress on the immune response and

    the outcome of pregnancy

    The hypothalamic-pituitary-adrenal (HPA) axis exerts

    an inhibitory effect on the female reproductive system

    when activated by stress. Corticotrophin releasing

    hormone (CRH) inhibits hypothalamic gonadotropin

    releasing hormone (GnRH) secretion, and glucocorti-

    coids inhibit pituitary luteinizing hormone and

    ovarian steroid hormones, estrogen and P.

    Psychosocial stress has been shown to alter cyto-

    kine production by peripheral lymphocytes of preg-

    nant women during the first trimester of pregnancy.

    Some of the pregnancy complications cannot be

    explained by either maternal or fetal pathologies.74–77

    Several epidemiological studies support the notion

    that the onset of miscarriages may be attributable to

    high levels of perceived stress. Repeated miscarriages

    can induce anxiety and even depression. Emotional

    stress, due to repeated pregnancy losses might also

    contribute to further miscarriages. However, to date,

    the unconfined acceptance of stress as a cause for

    pregnancy loss in everyday clinical practice is limited

    by contradictory observations.78,79 Such contradic-

    tory results may be explained by the diverse experi-

    mental design, such as correlating the temporal

    coincidence of stressful life events or self-reported

    stress perception to the onset of spontaneous

    abortion. Further, the lack of appropriate tools to

    evaluate stress perception is clearly a limitation in

    cohort studies. For humans, little physiological

    evidence exists in support of this hypothesis and

    thus, identification of risk factors for stress-triggered

    miscarriages requires further research.80–82

    Emerging evidence indicates that mediators of the

    HPA axis, such as CRH and the glucocorticoid corti-

    sol may serve as such stress indicators.83 High levels

    of glucocorticoids exert adverse effects on the uterus

    and fetus, and inhibit pituitary luteinizing hormone,

    and ovarian estrogen, and P secretion.84 Such inhibi-

    tory effects of stress hormones on the female

    reproductive system are responsible for the ‘hypo-

    thalamic’ amenorrhea of stress, and – as shown in

    mice – may also account for inadequate levels of

    P during pregnancy, subsequently resulting in spon-

    taneous abortion.84 The concept of stress-triggered

    inhibition of P is supported by experimental evi-

    dence from animal studies. Exposure to stress in the

    form of restraint85 or sound86 induces abortion in

    pregnant mice via a significant reduction in P levels,

    accompanied by reduced serum levels of P-induced

    blocking factor (PIBF) and diminished expression of

    PRs at the feto-maternal interface.87 Administration

    of the P derivative dydrogesterone increases levels of

    PIBF and restores the pregnancy-protective immune

    milieu in the mouse model of stress-triggered abor-

    tions, as well as in humans with threatened abor-

    tion.87–89 Such endocrine-immune cross talk is

    exceedingly dependent on a specific CD8+ T-cell

    population, since depletion of CD8 led to a termina-

    tion of the pregnancy protective effect of P substitu-

    tion in mice, whereby the precise phenotype of this

    specific, pregnancy-protective CD8 cell population,

    e.g. the co-expression of the ab or cd T-cell receptor,remains to be elucidated.88 The notion of decreased

    levels of P in response to stress could also be con-

    firmed in other mammalian species, such as in

    elks.90

    In addition to the ‘classical’ stress mediators, such

    as CRH, adrenocorticotropin (ACTH), cortisol or

    catecholamines,91 the neurotrophin nerve growth

    factor (NGF) or the neuropeptide Substance P are

    progressively recognized as a pivotal regulator of the

    ARCK ET AL.

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    272 Journal compilation ª 2007 Blackwell Munksgaard

  • stress response cascade.92–94 Thus, future research

    addressing the potential threat of such stress media-

    tors on progesterone production and pregnancy

    maintenance is needed.

    Endocrine-immune cross talk in Bovidae: insights

    into the immunological consequences of evolution

    of the epitheliochorial placenta.

    Besides mice and humans the Bovidae have been

    one of the most extensively studied clades in mam-

    malian reproduction; whereby examination of the

    specializations acquired by these animals during evo-

    lution provides insights into immunological adjust-

    ments to pregnancy. Such features are essential in

    reproduction in eutherian mammals and represent

    clade-specific solutions to the immunological prob-

    lem of viviparity.

    The Bovidae diverged as a separate family of peco-

    ran ruminants about 24–29 million years ago during

    the Late Oligocene epoch.95 While the basic pattern

    of reproduction in ruminants is similar to other

    mammals, there are distinct features including pla-

    cental anatomy. Ruminants possess an epitheliocho-

    rial placenta characterized by apposition of fetal and

    maternal tissues. Invasion of the maternal system

    either does not occur or is limited to migration of

    trophoblast cells into the maternal endometrial epi-

    thelium to form a syncytium (Fig. 1).96,97

    Evolutionary advantages conferred by an

    epitheliochorial placenta include more efficient

    transport of nutrients.96 In addition, fetal-maternal

    competition may be reduced in species with epithe-

    liochorial species because the mother has greater

    control over maternal blood flow to the placenta.96

    It is also possible that there are immunological

    consequences of epithelichorial placentation. As

    compared to species with invasive placenta, access of

    maternal leukocytes to fetal placental tissue is

    restricted physically by several cell layers and there

    may be reduced opportunity for pieces of trophoblast

    tissue to enter draining lymph nodes and peripheral

    circulation of the mother.

    Whether these differences actually confer

    increased immunological fitness for the placenta

    with respect to maternal immunological recognition

    is not known. In any case, the immunological rela-

    tionship between the conceptus and mother in Bovi-

    dae is similar in many ways to that for other

    mammals. Expression of MHC antigens on the tro-

    phoblast is largely down-regulated98,99 although, at

    least in the cow, there is limited expression of class I

    MHC molecules by trophoblast in later pregnancy.99

    Down-regulation of MHC antigen expression may be

    an important requirement for successful pregnancies:

    cloned bovine conceptuses, which experience high

    rates of fetal loss, can express aberrantly high

    levels of MHC class I protein associated with

    Fetal capillary

    Endothelium

    Maternal capillary

    Endothelium

    Fetal side Maternal side

    Connective

    tissueCon

    nect

    ive

    tissu

    e

    Fetal chorionic epithelium Maternal endometrial epithelium

    Epitheliochorial placenta

    Endotheliochorial placenta (Endo P) (Endo P)

    Hemochorial placenta

    Fig. 1 Comparative understanding of the

    placenta in different species: Placentas are

    variously classified, i.e. by their macroscopic

    appearance, or according to its intimacy of

    fetal-maternal contact. Here, the main placen-

    tal types have been described as epitheliocho-

    rial (three maternal layers and three fetal

    layers), endotheliochorial (one maternal layer,

    three fetal layers), hemochorial (no maternal

    layers; three fetal layers). (Note: placentas can

    also express a mosaic type). Hence, six cellu-

    lar layers that can potentially be between the

    fetal and maternal blood cells. Epitheliochorial

    (6-layer) placentas are common in pigs, cows,

    horses, and sheep. Endotheliochorial (4-layer)

    placentas are frequently found in dogs, cats,

    seals, and ferrets. Hemochorial placentas

    (where the maternal blood cells are in direct

    contact with the fetal chorion) are seen in

    humans, rats, and mice.114

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 273

  • increased accumulation of maternal lymphocytes in

    endometrial stroma.100

    Experiments from the sheep indicate that exten-

    sive remodeling of the leukocyte population in the

    uterus takes place during pregnancy. Macrophages

    accumulate in large numbers in the endometrial

    stroma during pregnancy101 and granulated cd-Tcells become abundant in luminal epithelium of the

    interplacentomal regions during mid- and late-preg-

    nancy.102,103 Non-granulated T cells in the glandular

    epithelium decline during pregnancy while numbers

    of these cells in luminal epithelium first decline and

    then return to levels seen in non-pregnant ewes.103

    It is not known whether there are changes in NK-

    cell populations in the endometrium during preg-

    nancy because of the paucity of immunological and

    functional assays for these cells in ruminants. Lim-

    ited evidence in sheep suggests that there is no large

    increase in endometrial NK-cell numbers in late

    pregnancy when compared with cyclic ewes.104 It

    may be that these cells, which play a crucial role in

    vascular remodeling in mice,105 are not important

    players in at least some species with epitheliochorial

    placentation because of differences in endometrial

    vascular architecture.

    Use of a unilaterally pregnant model in sheep

    (where pregnancy is surgically confined to one uter-

    ine horn) has revealed that accumulation of macro-

    phages is due to both systemic signals (numbers of

    cells in the non-pregnant uterine horn of the unilat-

    erally pregnant ewe higher than amounts in uteri of

    non-pregnant ewes) and locally produced signals

    (number of cells in the uterus of unilaterally ligated

    ewes higher in the pregnant horn than in the non-

    pregnant horn).102 Accumulation of cd T cells isa result of unidentified systemic signals.103 However,

    local placentally derived signals may be involved in

    activation of cd T cells. There was increased expres-sion of CD25 from cd T cells isolated from the preg-nant uterine horn of unilaterally pregnant ewes

    when compared with the non-pregnant horn.29

    As previously outlined, one of the key regulators

    of uterine immune function is P. In cattle, the pla-

    cental contribution to P synthesis is low throughout

    most of pregnancy and also low in sheep until day

    50 of pregnancy (gestation length = 147 days). Thus,

    for all or part of pregnancy, lymphocytes at the

    fetal-maternal interface are probably not exposed to

    the high concentrations required to inhibit lympho-

    cyte function.106 Administration of P to sheep can

    block tissue graft rejection in utero when injected to

    achieve concentrations in blood too low to directly

    inhibit lymphocyte proliferation.107,108 Thus, P regu-

    lates tissue rejection responses indirectly by inducing

    secretory molecules from the uterine endometrium

    that can regulate immune function.

    Interestingly, the major P-induced immunoregula-

    tory molecule in sheep is a member of the serine

    proteinase inhibitor family called uterine serpin.

    Ovine uterine serpin can block lymphocyte prolifera-

    tion in vitro in sheep109 and NK-cell-mediated abor-

    tion in vivo in mice.110 Cattle, goats, and pigs also

    secrete uterine serpin from the endometrium but its

    role in immune function in these species has not

    been documented. Strikingly, the gene for uterine

    serpin is not present in human or mouse as deter-

    mined by queries of genomic databases. The gene for

    uterine serpin arose early in mammalian evolu-

    tion111 and it may be that the gene has been retained

    only in species with epitheliochorial placentation.

    Local signals controlling macrophage accumulation

    and activation status of cd T cells have not beenidentified. The bovine placenta expresses non-classi-

    cal MHC antigens,112 and it may be that, as has been

    postulated for species with invasive placenta,113

    these molecules act to regulate macrophages and

    dendritic cells in the uterus to direct immune

    responses to favor conceptus survival.

    Conclusions

    Progesterone is critical for the establishment and the

    maintenance of pregnancy, both by its endocrine

    and immunological effects, as shown by a progester-

    one-dependent immunomodulation in maternal tol-

    erance of the fetus, e.g. via PIBF and uterine

    homing of NK cells and up-regulation of HLA-G

    gene expression in mice and ⁄ or humans. Adequatelevels of progesterone may be inhibited upon high

    stress perception, followed by reduced serum levels

    of PIBF and diminished expression of progesterone

    receptors at the feto-maternal interface. These effects

    are corrected by progesterone supplementation.

    Some divergence in the pattern of endocrine-

    immunological cross talk is present in Bovidae, such

    as the lack of placental progesterone synthesis or the

    presence of uterine serpin, a progesterone-induced

    endometrial protein, which may mediate the immu-

    nosuppressive effects of progesterone in sheep. By

    reason of their evolutionary conservation, these

    may be essential features of vivaparity in eutherian

    mammals.

    ARCK ET AL.

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    274 Journal compilation ª 2007 Blackwell Munksgaard

  • Acknowledgements

    P Arck, B Mulac Jericevic, M-P Piccinni and J Sze-

    keres-Bartho are members of the European Network

    of Excellence on Embryo Implantation Control, sup-

    ported by EU Contract No. 512040.

    References

    1 Stites DP, Siiteri PK: Steroids as immunosuppressants

    in pregnancy. Immunol Rev 1983; 75:117–138.

    2 Miyaura H, Iwata M: Direct and indirect inhibition

    of Th1 development by progesterone and

    glucocorticoids. J Immunol 2002; 168:1087–1094.

    3 Tibbetts TA, DeMayo F, Rich S, Conneely OM,

    O’Malley BW: Progesterone receptors in the thymus

    are required for thymic involution during pregnancy

    and for normal fertility. Proc Natl Acad Sci USA 1999;

    96:12021–12026.

    4 Piccinni M-P, Giudizi MG, Biagiotti R, Beloni L,

    Giannarini L, Sampognaro S, Parronchi P, Manetti R,

    Livi C, Romagnani S, Maggi E: Progesterone favors

    the development of human T helper cells producing

    Th2-type cytokines and promotes both IL-4

    production and membrane CD30 expression in

    established Th1 cells clones. J Immunol 1995;

    155:128–133.

    5 Szekeres-Bartho J, Kilar F, Falkay G, Csernus V,

    Torok A, Pacsa AS: Progesterone-treated

    lymphocytes of healthy pregnant women release a

    factor inhibiting cytotoxicity and prostaglandin

    synthesis. Am J Reprod Immunol Microbiol 1985;

    9:15–19.

    6 Szekeres-Bartho J: Immunological relationship

    between the mother and the fetus. Int Rev Immunol

    2002; 21:471–495.

    7 Kayisli UA, Guzeloglu-Kayisli O, Arici A: Endocrine-

    immune interactions in human endometrium. Ann

    N Y Acad Sci 2004; 1034:50–63.

    8 Dosiou C, Giudice LC: Natural killer cells in pregnancy

    and recurrent pregnancy loss: endocrine and

    immunologic perspectives. Endocr Rev 2005; 26:44–62.

    9 Li X, Lonard DM, O’Malley BW: A contemporary

    understanding of progesterone receptor function.

    Mech Ageing Dev 2004; 125:669–678.

    10 Mulac-Jericevic B, Conneely OM: Reproductive

    tissue selective actions of progesterone receptors.

    Reproduction 2004; 128:139–146.

    11 Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes

    AR, Montgomery CA Jr, Shyamala G, Conneely OM,

    O’Malley BW: Mice lacking progesterone receptor

    exhibit pleiotropic reproductive abnormalities. Genes

    Dev 1995; 9:2266–2278.

    12 Tibbetts TA, Conneely OM, O’Malley BW:

    Progesterone via its receptor antagonizes the pro-

    inflammatory activity of estrogen in the mouse

    uterus. Biol Reprod 1999; 60:1158–1165.

    13 Mulac-Jericevic B, Mullinax RA, DeMayo FJ, Lydon

    JP, Conneely OM: Subgroup of reproductive

    functions of progesterone mediated by progesterone

    receptor-B isoform. Science 2000; 289:1751–1754.

    14 Mulac-Jericevic B, Lydon JP, DeMayo FJ, Conneely

    OM: Defective mammary gland morphogenesis in

    mice lacking the progesterone receptor B

    isoform. Proc Natl Acad Sci U S A 2003;

    100:9744–9749.

    15 Fernandez-Valdivia R, Mukherjee A, Mulac-Jericevic

    B, Conneely OM, DeMayo FJ, Amato P, Lydon JP:

    Revealing progesterone’s role in uterine and

    mammary gland biology: insights from the mouse.

    Semin Reprod Med 2005; 23:22–37.

    16 Choe YS, Shim C, Choi D, Lee CS, Lee KK, Kim K:

    Expression of galectin-1 mRNA in the mouse uterus

    is under the control of ovarian steroids during

    blastocyst implantation. Mol Reprod Dev 1997;

    48:261–266.

    17 Yao MW, Lim H, Schust DJ, Choe SE, Farago A,

    Ding Y, Michaud S, Church GM, Maas RL: Gene

    expression profiling reveals progesterone-mediated

    cell cycle and immunoregulatory roles of Hoxa-10 in

    the preimplantation uterus. Mol Endocrinol 2003;

    17:610–627.

    18 Croy BA, van den Heuvel MJ, Borzychowski AM,

    Tayade C: Uterine natural killer cells: a specialized

    differentiation regulated by ovarian hormones.

    Immunol Rev 2006; 214:161–185.

    19 Rahman MA, Li M, Li P, Wang H, Dey SK, Das SK:

    Hoxa-10 deficiency alters region-specific gene

    expression and perturbs differentiation of natural

    killer cells during decidualization. Dev Biol 2006;

    290:105–117.

    20 Jeschke U, Richter DU, Hammer A, Briese V, Friese

    K, Karsten U: Expression of the Thomsen-

    Friedenreich antigen and of its putative carrier

    protein mucin 1 in the human placenta and in

    trophoblast cells in vitro. Histochem Cell Biol 2002;

    117:219–226.

    21 Aplin JD, Meseguer M, Simon C, Ortiz ME, Croxatto

    H, Jones CJ: MUC1, glycans and the cell-surface

    barrier to embryo implantation. Biochem Soc Trans

    2001; 29:153–156.

    22 Brayman MJ, Julian J, Mulac-Jericevic B, Conneely

    OM, Edwards DP, Carson DD: Progesterone receptor

    isoforms A and B differentially regulate MUC1

    expression in uterine epithelial cells. Mol Endocrinol

    2006; 20:2278–2291.

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 275

  • 23 Lee K, Jeong J, Kwak I, Yu CT, Lanske B, Soegiarto

    DW, Toftgard R, Tsai MJ, Tsai S, Lydon JP, DeMayo

    FJ: Indian hedgehog is a major mediator of

    progesterone signaling in the mouse uterus. Nat

    Genet 2006; 38:1204–1209.

    24 Liu AX, Jin F, Zhang WW, Zhou TH, Zhou CY, Yao

    WM, Qian YL, Huang HF: Proteomic analysis on the

    alteration of protein expression in the placental

    villous tissue of early pregnancy loss. Biol Reprod

    2006; 75:414–420.

    25 Mesiano S: Myometrial progesterone responsiveness.

    Semin Reprod Med 2007; 25:5–13.

    26 Giudice LC: Application of functional genomics to

    primate endometrium: insights into biological

    processes. Reprod Biol Endocrinol 2006; 4(Suppl. 1): S4.

    27 Horcajadas JA, Pellicer A, Simon C: Wide genomic

    analysis of human endometrial receptivity: new

    times, new opportunities. Hum Reprod Update 2007;

    13:77–86.

    28 Wang H, Dey SK: Roadmap to embryo implantation:

    clues from mouse models. Nat Rev Genet 2006;

    7:185–199.

    29 Liu WJ, Gottshall SL, Hansen PJ: Increased

    expression of cell surface marker on endometrial g ⁄ dT cell receptor intraepithelial lymphocytes induced

    by the local presence of the sheep conceptus.

    Am J Reprod Immunol 1997; 37:199–205.

    30 Meeusen E, Fox A, Brandon M, Lee CS: Activation

    of uterine intraepithelial gamma delta T cell receptor

    positive lymphocytes during pregnancy.

    Eur J Immunol 1993; 23:1112–1117.

    31 Szekeres-Bartho J, Barakonyi A, Miko E, Polgar B,

    Palkovics T: The role of c ⁄ d T cells in the feto-maternal relationship. Semin Immunol 2001;

    13:229–233.

    32 Kämmerer U, Schoppet M, McLellan AD, Kapp M,

    Huppertz HI, Kampgen E, Dietl J: Human decidua

    contains potent immunostimulary CD83+ dendritic

    cells. Am J Pathol 2000; 157:159–169.

    33 Gardner L, Moffett A: Dendritic cells in the human

    decidua. Biol Reprod 2003; 69:1438–1446.

    34 Blois SM, Tometten M, Kandil J, Hagen E, Klapp BF,

    Margni RA, Arck PC: ICAM-1 ⁄ LFA-1 cross talk is aproximate mediator capable of disrupting immune

    integration and tolerance mechanism at the feto-

    maternal interface in murine pregnancies. J Immunol

    2005; 174:1820–1829.

    35 Moretta A, Marcenaro E, Sivori S, Della Chiesa M,

    Vitale M, Moretta L: Early liaisons between cells of

    the innate immune system in inflamed peripheral

    tissues. Trends Immunol 2005; 26:668–675.

    36 Kämmerer U: Antigen-presenting cells in the

    decidua. Chem Immunol Allergy 2005; 89:96–104.

    37 Blois SM, Alba Soto CD, Tometten M, Klapp BF,

    Margni RA, Arck PC: Lineage, maturity, and

    phenotype of uterine murine dendritic cells

    throughout gestation indicate a protective role in

    maintaining pregnancy. Biol Reprod 2004;

    70:1018–1023.

    38 Zhu XY, Zhou YH, Wang MY, Jin LP, Yuan MM, Li

    DJ: Blockade of CD86 signaling facilitates a Th2 bias

    at the maternal-fetal interface and expands peripheral

    CD4+CD25+ regulatory T cells to rescue abortion-

    prone fetuses. Biol Reprod 2005; 72:338–345.

    39 Butts CL, Shukair SA, Duncan KM, Bowers E, Horn

    C, Belyayskaya E, Tonelli L, Sternbers EM:

    Progesterone inhibits mature rat dendritic cells in a

    receptor-mediated fashion. Int Immunol 2007;

    19:287–296.

    40 Szekeres-Bartho J, Szekeres GY, Debre P, Autran B,

    Chaouat G: Reactivity of lymphocytes to a

    progesterone receptor-specific monoclonal antibody.

    Cell Immunol 1990; 125:273–283.

    41 Chiu L, Nishimura M, Ishi Y, Nieda M, Maeshima M,

    Takedani Y, Tadokoro K, Juji T: Enhancement of the

    expression of progesterone receptor on progesterone

    -treated lymphocytes after immunotherapy in

    unexplained recurrent spontaneous abortion.

    Am J Reprod Immunol 1996; 35:552–557.

    42 van den Heuvel M, McBey B-A, Hahnel AC, Croy

    BA: An analysis of the uterine lymphocyte-derived

    hybridoma cell line GWM 1-2 for expression of

    receptors for estrogen, progesterone and interleukin

    2. J Reprod Immunol 1996; 31:37–50.

    43 Roussev RG, Higgins NG, McIntyre JA: Phenotypic

    characterization of normal human placental

    mononuclear cells. J Reprod Immunol 1993; 25:15–29.

    44 Chien EJ, Liao CF, Chang CP, Pu HF, Lu LM, Shie

    MC, Hsieh DJ, Hsu MT: The non-genomic effects on

    Na(+) ⁄ H(+)-exchange 1 by progesterone and20alpha-hydroxyprogesterone in human T cells.

    J Cell Physiol 2007; 211:544–550.

    45 Polgar B, Kispal GY, Lachmann M, Paar C, Nagy E,

    Csere P, Miko E, Szereday L, Varga P, Szekeres-

    Bartho J: Molecular cloning and immunological

    characterization of a novel cDNA coding for PIBF.

    J Immunol 2003; 171:5956–5963.

    46 Polgár B, Nagy E, Mikó É, Varga P, Szekeres-Barthó

    J: Urinary PIBF (Progesterone Induced Blocking

    Factor) concentration is related to pregnancy

    outcome. Biol Reprod 2004; 71:1699–1705.

    47 Kozma N, Halasz M, Polgar B, Poehlmann TG,

    Markert UR, Palkovics T, Keszei M, Kiss K,

    Szeberenyi J, Par G, Grama L, Szekeres-Bartho J:

    PIBF activates STAT6 via binding to a novel IL-4

    receptor. J Immunol 2006; 176:819–826.

    ARCK ET AL.

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    276 Journal compilation ª 2007 Blackwell Munksgaard

  • 48 Szekeres-Bartho J, Wegmann TG: A progesterne-

    dependent immunomodulatory protein alters the

    Th1 ⁄ Th2 balance. J Reprod Immunol 1996; 31:81–95.49 Szekeres-Bartho J, Faust ZS, Varga P, Szereday L,

    Kelemen K: The immunological pregnancy

    protective effect of progesterone is manifested via

    controlling cytokine production. Am J Reprod

    Immunol 1996; 35:348–351.

    50 Szekeres-Bartho J, Par G, Dombay GY, Smart YC,

    Volgyi Z: The anti-abortive effect of PIBF in mice is

    manifested by modulating NK activity. Cell Immunol

    1997; 177:194–199.

    51 King A, Burrows T, Verma S, Hiby S, Loke YW:

    Human uterine lymphocytes. Hum Reprod Update

    1998; 4:480–485.

    52 Hanna J, Goldman-Wohl D, Hamani Y, Avraham I,

    Greenfield C, Nathanson-Yaron S, Prus D, Cohen-

    Daniel L, Arnon TI, Manaster I, Gazit R, Yutkin V,

    Beaharroch D, Porgador A, Keshet E, Yagel S,

    Mandelboim O: Decidual NK cells regulate key

    developmental processes at the human

    fetal-maternal interface. Nat Med 2006; 12:

    1065–1074.

    53 Henderson TA, Saunders PT, Moffet-King A, Groome

    NP, Chritchley HO: Steroid receptor expression in

    uterine natural killer cells. J Clin Endocrinol Metab

    2003; 88:440–449.

    54 Sentman CL, Meadows SK, Wira CR, Eriksson M:

    Recruitment of uterine NK cells: induction of CXC

    Chemokine ligands 10 and 11 inhuman

    endometrium by estradiol and progesterone.

    J Immunol 2004; 173:6760–6766.

    55 Crncic TB, Laskarin G, Frankovic KJ, Tokmadzic VS,

    Strobo N, Bedenicki I, Le Bouteiller P, Tabiasco J,

    Rukavina D: Early pregnancy decidual lymphocytes

    beside perforin use Fas ligand (FasL) mediated

    cytotoxicity. J Reprod Immunol 2007; 73:108–117.

    56 Yie SM, Xiao R, Librach CL: Progesterone regulates

    HLA-G gene expression through a novel

    progesterone response element. Hum Reprod 2006;

    21:2538–2544.

    57 Mosmann TR, Coffman RL: Th1 and Th2 cells:

    different patterns of lymphokine secretion lead to

    different functional properties. Annu Rev Immunol

    1989; 7:145–173.

    58 Romagnani S: Human Th1 and Th2: doubt no more.

    Immunol Today 1991; 12:256–257.

    59 Weiner LH: Induction and mechanism of action of

    transforming growth factor beta secreting Th3

    regulatory cells. Immunol Rev 2001; 182:207–214.

    60 Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-

    Joannopoulos K, Collins M, Fouser LA: Interleukin

    (IL)-22 and IL-17 are coexpressed by Th17 cells and

    cooperatively enhance expression of antimicrobial

    peptides. J Exp Med 2006; 203:2271–2279.

    61 Huber S, Schramm C, Lehr HA, Mann A, Schmitt S,

    Becker C, Protschka M, Galle PR, Neurath MF,

    Blessing M: Cutting edge: TGF-beta signaling is

    required for the in vivo expansion and

    immunosuppressive capacity of regulatory

    CD4+CD25+ T cells. J Immunol 2004; 173:6526–6531.

    62 Maggi E, Parronchi P, Manetti R, Simonelli C,

    Piccinni M-P, Santoni-Rugiu F, De Carli M, Ricci

    M, Romagnani S: Reciprocal regulatory role of

    IFN-c and IL-4 on the in vitro development ofhuman Th1 and Th2 clones. J Immunol 1992;

    148:2142–2147.

    63 Manetti R, Parrochi P, Giudizi MG, Piccinni M-P,

    Maggi E, Trinchieri G, Romagnani S: Natural killer

    cell stimulatory factor (interleukin-12) induces

    T helper type 1 (Th1)-specific immune responses and

    inhibits the development of IL-4-producing Th cells.

    J Exp Med 1993; 177:1199–1204.

    64 Daynes RA, Meikle AW, Araneo BA: Locally active

    steroid hormones may facilitate

    compartmentalization of immunity by regulating the

    types of lymphokines produced by helper T cells. Res

    Immunol 1991; 142:40–45.

    65 Rook GAW, Hernandez-Pando R, Lightman SL:

    Hormones, peripherally activated prohormones and

    regulation of the Th1 ⁄ Th2 balance. Immunol Today1994; 13:301–303.

    66 Vacca A, Martinotti S, Screpanti I, Maroder M, Felli

    MP, Farina AR, Gismondi A, Santoni A, Frati L,

    Gulino A: Transcriptional regulation of the

    interleukin 2 gene by glucocorticoid hormones.

    J Biol Chem 1990; 265:8075–8080.

    67 Piccinni M-P, Bani D, Beloni L, Manuelli C, Mavilia

    C, Vocioni F, Bigazzi M, Sacchi TB, Romagnani S,

    Maggi E: Relaxin favors the development of

    activated human T cells into Th1-like effectors.

    Eur J Immunol 1999; 29:2241–2247.

    68 Wang Y, Campbell HD, Young IG: Sex hormones

    and dexamethasone modulate interleukin-5 gene

    expression in T lymphocytes. J Steroid Biochem Mol

    Biol 1993; 44:203–210.

    69 Piccinni M-P, Beloni L, Livi C, Maggi E, Scarselli GF,

    Romagnani S: Defective production of both leukemia

    inhibitory factor and type 2 T-helper cytokines by

    decidual T cells in unexplained recurrent abortions.

    Nat Med 1998; 4:1020–1024.

    70 Piccinni M-P, Scaletti C, Vultaggio A, Maggi E,

    Romagnani S: Defective production of LIF, M-CSF

    and Th2-type cytokines by T cells at fetomaternal

    interface is associated with pregnancy loss. J Reprod

    Immunol 2001; 52:35–43.

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 277

  • 71 Wegmann TG, Lin L, Guilbert L, Mosmann TR:

    Bidirectional cytokine interactions in the maternal-

    fetal relationship: is successful pregnancy a

    Th2 phenomenon?. Immunol Today 1993; 14:

    353–356.

    72 Delassus S, Coutinho GC, Saucier C, Darche S,

    Kourilsky P: Differential cytokine expression in

    maternal blood and placenta during murine

    gestation. J Immunol 1994; 152:2411–2420.

    73 Piccinni MP: T cells in normal pregnancy and

    recurrent pregnancy loss. Reprod Biomed Online 2006;

    13:840–844.

    74 Lanasa MC, Hogge WA, Kubik CJ, Ness RB, Harger

    J, Nagel T, Prosen T, Markovic N, Hoffman EP: A

    novel X chromosome-linked genetic cause of

    recurrent spontaneous abortion. Am J Obstet Gynecol

    2001; 185:563–568.

    75 Rubio C, Gil-Salom M, Simon C, Vidal F, Rodrigo L,

    Minguez Y, Remohi J, Pellicer A: Incidence of sperm

    chromosomal abnormalities in a risk population:

    relationship with sperm quality and ICSI outcome.

    Hum Reprod 2001; 16:2084–2092.

    76 Arck P, Knackstedt M, Blois S: Neuroendocrine

    immune circuitry challenging pregnancy

    maintenance and fetal health. J Reprod Med

    Endocrinol 2006; 2:98–102.

    77 Arck PC, Rose M, Hertwig K, Hagen E, Hildebrandt

    M, Klapp BF: Stress and immune mediators in

    miscarriage. Hum Reprod 2001; 16:1505–1511.

    78 Fenster L, Schaefer C, Mathur A, Hiatt RA, Pieper C,

    Hubbard AE, Von Behren J, Swan SH: Psychologic

    stress in the workplace and spontaneous abortion.

    Am J Epidemiol 1995; 142:1176–1183.

    79 Nepomnaschy PA, Welch KB, McConnell DS, Low

    BS, Strassmann BI, England BG: Cortisol levels and

    very early pregnancy loss in humans. Proc Natl Acad

    Sci U S A 2006; 103:3938–3942.

    80 Neugebauer R, Kline J, Stein Z, Shrout P, Warburton

    D, Susser M: Association of stressful life events with

    chromosomally normal spontaneous abortion.

    Am J Epidemiol 1996; 143:588–596.

    81 Stray-Pedersen BS, Stray-Pedersen S: Etiologic

    Factors and susequent reproductive performance in

    195 couples with a prior history of habitual abortion.

    Am J Obstet Gynecol 1984; 148:140–146.

    82 Nelson DB, Grisso JA, Joffe MM, Brensinger C,

    Shaw L, Datner E: Does stress influence early

    pregnancy loss?. Ann Epidemiol 2003; 13:223–229.

    83 Milad MP, Klock SC, Moses S, Chatterton R: Stress

    and anxiety do not result in pregnancy wastage.

    Hum Reprod 1998; 13:2296–2300.

    84 Magiakou MA, Mastorakos G, Webster E, Chrousos

    GP: The hypothalamic-pituitary-adrenal axis and the

    female reproductive system. Ann N Y Acad Sci 1997;

    816:42–56.

    85 Wiebold JL, Stanfield PH, Becker WC, Hillers JK:

    The effect of restraint stress in early pregnancy in

    mice. J Reprod Fertil 1986; 78:185–192.

    86 Joachim R, Zenclussen AC, Polgar B, Douglas AJ,

    Fest S, Knackstedt M, Klapp BF, Arck PC: The

    progesterone derivative dydrogesterone abrogates

    murine stress-triggered abortion by inducing a Th2

    biased local immune response. Steroids 2003;

    68:931–940.

    87 Blois SM, Joachim R, Kandil J, Margni R, Tometten

    M, Klapp BF, Arck PC: Depletion of CD8+ cells

    abolishes the pregnancy protective effect of

    progesterone substitution with dydrogesterone in

    mice by altering the Th1 ⁄ Th2 cytokine profile.J Immunol 2004; 172:5893–5899.

    88 Kalinka J, Szekeres-Bartho J: The impact of

    dydrogesterone supplementation on hormonal

    profile and progesterone-induced blocking factor

    concentrations in women with threatened abortion.

    Am J Reprod Immunol 2005; 53:166–171.

    89 Creel S, Christianson D, Liley S, Winnie JA:

    Predation risk affects reproductive physiology and

    demography of elk. Science 2007; 315:960.

    90 Benschop RJ, Rodriguez-Feuerhahn M, Schedlowski

    M: Catecholamine-induced leukocytosis: early

    observations, current research, and future directions.

    Brain Behav Immun 1996; 10:77–91.

    91 Aloe L, Alleva E, Fiore M: Stress and nerve growth

    factor: findings in animal models and humans.

    Pharmacol Biochem Behav 2002; 73:159–166.

    92 Tometten M, Klapp BF, Joachim R, Fest S,

    Zenclussen AC, Peters EM, Hertwig K, Arck PC:

    Nerve growth factor and its functional receptor TrkA

    are up-regulated in murine decidual tissue of stress-

    triggered and substance P-mediated abortion.

    Am J Reprod Immunol 2004; 51:86–93.

    93 Tometten M, Blois S, Kuhlmei A, Stretz A, Klapp BF,

    Arck PC: Nerve growth factor translates stress

    response and subsequent murine abortion via

    adhesion molecule-dependent pathways. Biol Reprod

    2006; 74:674–683.

    94 Joachim RA, Hildebrandt M, Oder J, Klapp BF, Arck

    PC: Murine stress-triggered abortion is mediated by

    increase of CD8+ TNF-alpha+ decidual cells via

    substance P. Am J Reprod Immunol 2001; 45:303–309.

    95 Hassanin A, Douzery EJ: Molecular and

    morphological phylogenies of ruminantia and the

    alternative position of the moschidae. Syst Biol 2003;

    52:206–228.

    96 Wildman DE, Chen C, Erez O, Grossman LI, Goodman

    M, Romero R: Evolution of the mammalian placenta

    ARCK ET AL.

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    278 Journal compilation ª 2007 Blackwell Munksgaard

  • revealed by phylogenetic analysis. Proc Natl Acad Sci U

    S A 2006; 103:3203–3208.

    97 Vogel P: The current molecular phylogeny of

    Eutherian mammals challenges previous

    interpretations of placental evolution. Placenta 2005;

    26:591–596.

    98 Gogolin-Ewens KJ, Lee CS, Mercer WR, Brandon

    MR: Site-directed differences in the immune

    response to the fetus. Immunology 1989; 66:312–317.

    99 Davies CJ, Hill JR, Edwards JL, Schrick FN, Fisher PJ,

    Eldridge JA, Schlafer DH: Major histocompatibility

    antigen expression on the bovine placenta: its

    relationship to abnormal pregnancies and retained

    placenta. Anim Reprod Sci 2004; 82–83:267–280.

    100 Hill JR, Schlafer DH, Fisher PJ, Davies CJ: Abnormal

    expression of trophoblast major histocompatibility

    complex class I antigens in cloned bovine pregnancies

    is associated with a pronounced endometrial

    lymphocytic response. Biol Reprod 2002; 67:55–63.

    101 Tekin S, Hansen PJ: Regulation of numbers of

    macrophages in the endometrium of the sheep by

    systemic effects of pregnancy, local presence of the

    conceptus, and progesterone. Am J Reprod Immunol

    2004; 51:56–62.

    102 Lee CS, Meeusen E, Gogolin-Ewens K, Brandon MR:

    Quantitative and qualitative changes in the

    intraepithelial lymphocyte population in the uterus

    of nonpregnant and pregnant sheep. Am J Reprod

    Immunol 1992; 28:90–96.

    103 Majewski AM, Tekin S, Hansen PJ: Local versus

    systemic control of numbers of endometrial T-cells

    during pregnancy in sheep. Immunology 2001;

    102:317–322.

    104 Tekin S, Hansen PJ: Lymphocyte-mediated lysis of

    sheep chorion: susceptibility of chorionic cells to

    third-party and maternal cytotoxic lymphocytes and

    presence of cells in the endometrium exhibiting

    cytotoxicity toward natural-killer cell targets.

    Theriogenology 2003; 59:787–800.

    105 Leonard S, Murrant C, Tayade C, van den Heuvel M,

    Watering R, Croy BA: Mechanisms regulating

    immune cell contributions to spiral artery

    modification – facts and hypotheses – a review.

    Placenta 2006; 27(Suppl. A): S40–S46.

    106 Monterroso VH, Hansen PJ: Regulation of bovine

    and ovine lymphocyte proliferation by progesterone:

    modulation by steroid receptor antagonists and

    physiological status. Acta Endocrinol 1993; 129:

    532–535.

    107 Majewski AC, Hansen PJ: Progesterone promotes

    survival of xenogeneic transplants in the sheep

    uterus. Horm Res 2002; 58:128–135.

    108 Padua MB, Tekin S, Spencer TE, Hansen PJ: Actions

    of progesterone on uterine immunosuppression and

    endometrial gland development in the uterine gland

    knockout (UGKO) ewe. Mol Reprod Dev 2005;

    71:347–357.

    109 Peltier MR, Liu W-J, Hansen PJ: Regulation of

    lymphocyte proliferation by uterine serpin:

    interleukin-2 mRNA production, CD25 expression

    and responsiveness to interleukin-2. Proc Soc Exp Biol

    Med 2000; 223:75–81.

    110 Liu W-J, Hansen PJ: Effect of the progesterone-

    induced serpin-like proteins of the sheep

    endometrium on natural-killer cell activity in

    sheep and mice. Biol Reprod 1993; 49:

    1008–1014.

    111 Peltier MR, Raley LC, Liberles DA, Benner SA,

    Hansen PJ: Evolutionary history of Evolutionary

    history of the uterine serpins. J Exp Zool 2000;

    288:165–174.

    112 Davies CJ, Eldridge JA, Fisher PJ, Schlafer DH:

    Evidence for expression of both classical and non-

    classical major histocompatibility complex class I

    genes in bovine trophoblast cells. Am J Reprod

    Immunol 2006; 55:188–200.

    113 Hunt JS, Petroff MG, McIntire RH, Ober C: HLA-G

    and immune tolerance in pregnancy. FASEB J 2005;

    19:681–693.

    114 Behringer RR, Eakin GS, Renfree MB: Mammalian

    diversity: gametes, embryos and reproduction. Reprod

    Fertil Dev 2006; 18:99–107.

    PROGESTERONE DURING PREGNANCY

    American Journal of Reproductive Immunology 58 (2007) 268–279 ª 2007 The Authors

    Journal compilation ª 2007 Blackwell Munksgaard 279